Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Trends Immunol ; 43(2): 148-162, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35033428

RESUMEN

Ubc13-catalyzed K63 ubiquitination is a major control point for immune signaling. Recent evidence has shown that the control of multiple immune functions, including chronic inflammation, pathogen responses, lymphocyte activation, and regulatory signaling, is altered by K63 ubiquitination. In this review, we detail the novel cellular sensors that are dependent on K63 ubiquitination for their function in the immune signaling network. Many pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can target K63 ubiquitination to inhibit pathogen immune responses; we describe novel details of the pathways involved and summarize recent clinically relevant SARS-CoV-2-specific responses. We also discuss recent evidence that regulatory T cell (Treg) versus T helper (TH) 1 and TH17 cell subset regulation might involve K63 ubiquitination. Knowledge gaps that merit future investigation and clinically relevant pathways are also addressed.


Asunto(s)
COVID-19 , Lisina , Humanos , Lisina/metabolismo , SARS-CoV-2 , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
2.
Mov Disord ; 38(6): 1056-1067, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37066491

RESUMEN

BACKGROUND: The intercellular transmission of pathogenic proteins plays a crucial role in the progression of neurodegenerative diseases. Previous research has shown that the neuronal uptake of such proteins is activity-dependent; however, the detailed mechanisms underlying activity-dependent α-synuclein transmission in Parkinson's disease remain unclear. OBJECTIVE: To examine whether α-synuclein transmission is affected by Ca2+ -calmodulin-calcineurin signaling in cultured cells and mouse models of Parkinson's disease. METHODS: Mouse primary hippocampal neurons were used to examine the effects of the modulation of Ca2+ -calmodulin-calcineurin signaling on the neuronal uptake of α-synuclein preformed fibrils. The effects of modulating Ca2+ -calmodulin-calcineurin signaling on the development of α-synuclein pathology were examined using a mouse model injected with α-synuclein preformed fibrils. RESULTS: Modulation of Ca2+ -calmodulin-calcineurin signaling by inhibiting voltage-gated Ca2+ channels, calmodulin, and calcineurin blocked the neuronal uptake of α-synuclein preformed fibrils via macropinocytosis. Different subtypes of voltage-gated Ca2+ channel differentially contributed to the neuronal uptake of α-synuclein preformed fibrils. In wild-type mice inoculated with α-synuclein preformed fibrils, we found that inhibiting calcineurin ameliorated the development of α-synuclein pathology. CONCLUSION: Our data suggest that Ca2+ -calmodulin-calcineurin signaling modulates α-synuclein transmission and has potential as a therapeutic target for Parkinson's disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Humanos , Animales , Ratones , alfa-Sinucleína/metabolismo , Enfermedad de Parkinson/patología , Calmodulina/metabolismo , Calcineurina/metabolismo , Neuronas/metabolismo
3.
J Hum Genet ; 67(7): 393-397, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35087201

RESUMEN

Paucity of interlobular bile ducts (PILBD) is a heterogeneous disorder classified into two categories, syndromic and non-syndromic bile duct paucity. Syndromic PILBD is characterized by the presence of clinical manifestations of Alagille syndrome. Non-syndromic PILBD is caused by multiple diseases, such as metabolic and genetic disorders, infectious diseases, and inflammatory and immune disorders. We evaluated a family with a dominantly inherited PILBD, who presented with cholestasis at 1-2 months of age but spontaneously improved by 1 year of age. Next-generation sequencing analysis revealed a heterozygous CACYBP/SIP p.E177Q pathogenic variant. Calcyclin-binding protein and Siah1 interacting protein (CACYBP/SIP) form a ubiquitin ligase complex and induce proteasomal degradation of non-phosphorylated ß-catenin. Immunohistochemical analysis revealed a slight decrease in CACYBP and ß-catenin levels in the liver of patients in early infancy, which almost normalized by 13 months of age. The CACYBP/SIP p.E177Q pathogenic variant may form a more active or stable ubiquitin ligase complex that enhances the degradation of ß-catenin and delays the maturation of intrahepatic bile ducts. Our findings indicate that accurate regulation of the ß-catenin concentration is essential for the development of intrahepatic bile ducts and CACYBP/SIP pathogenic variant is a novel cause of PILDB.


Asunto(s)
Síndrome de Alagille , Proteínas de Unión al Calcio , beta Catenina , Conductos Biliares Intrahepáticos/metabolismo , Proteínas de Unión al Calcio/genética , Humanos , Lactante , Recién Nacido , Ubiquitina-Proteína Ligasas , beta Catenina/metabolismo
4.
Proc Natl Acad Sci U S A ; 116(27): 13404-13413, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31213539

RESUMEN

BRUCE/Apollon is a membrane-associated inhibitor of apoptosis protein that is essential for viability and has ubiquitin-conjugating activity. On initiation of apoptosis, the ubiquitin ligase Nrdp1/RNF41 promotes proteasomal degradation of BRUCE. Here we demonstrate that BRUCE together with the proteasome activator PA28γ causes proteasomal degradation of LC3-I and thus inhibits autophagy. LC3-I on the phagophore membrane is conjugated to phosphatidylethanolamine to form LC3-II, which is required for the formation of autophagosomes and selective recruitment of substrates. SIP/CacyBP is a ubiquitination-related protein that is highly expressed in neurons and various tumors. Under normal conditions, SIP inhibits the ubiquitination and degradation of BRUCE, probably by blocking the binding of Nrdp1 to BRUCE. On DNA damage by topoisomerase inhibitors, Nrdp1 causes monoubiquitination of SIP and thus promotes apoptosis. However, on starvation, SIP together with Rab8 enhances the translocation of BRUCE into the recycling endosome, formation of autophagosomes, and degradation of BRUCE by optineurin-mediated autophagy. Accordingly, deletion of SIP in cultured cells reduces the autophagic degradation of damaged mitochondria and cytosolic protein aggregates. Thus, by stimulating proteasomal degradation of LC3-I, BRUCE also inhibits autophagy. Conversely, SIP promotes autophagy by blocking BRUCE-dependent degradation of LC3-I and by enhancing autophagosome formation and autophagic destruction of BRUCE. These actions of BRUCE and SIP represent mechanisms that link the regulation of autophagy and apoptosis under different conditions.


Asunto(s)
Autofagia , Proteínas de Unión al Calcio/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Apoptosis , Autofagosomas/metabolismo , Daño del ADN , Fibroblastos , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Ubiquitinación
5.
Mov Disord ; 36(7): 1554-1564, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33813737

RESUMEN

BACKGROUND: The intercellular transmission of pathogenic proteins plays a key role in the clinicopathological progression of neurodegenerative diseases. Previous studies have demonstrated that this uptake and release process is regulated by neuronal activity. OBJECTIVE: The objective of this study was to examine the effect of perampanel, an antiepileptic drug, on α-synuclein transmission in cultured cells and mouse models of Parkinson's disease. METHODS: Mouse primary hippocampal neurons were transduced with α-synuclein preformed fibrils to examine the effect of perampanel on the development of α-synuclein pathology and its mechanisms of action. An α-synuclein preformed fibril-injected mouse model was used to validate the effect of oral administration of perampanel on the α-synuclein pathology in vivo. RESULTS: Perampanel inhibited the development of α-synuclein pathology in mouse hippocampal neurons transduced with α-synuclein preformed fibrils. Interestingly, perampanel blocked the neuronal uptake of α-synuclein preformed fibrils by inhibiting macropinocytosis in a neuronal activity-dependent manner. We confirmed that oral administration of perampanel ameliorated the development of α-synuclein pathology in wild-type mice inoculated with α-synuclein preformed fibrils. CONCLUSION: Modulation of neuronal activity could be a promising therapeutic target for Parkinson's disease, and perampanel could be a novel disease-modifying drug for Parkinson's disease. © 2021 International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , Ratones , Nitrilos , Enfermedad de Parkinson/tratamiento farmacológico , Piridonas/farmacología , alfa-Sinucleína/genética
6.
Brain ; 143(1): 249-265, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31816026

RESUMEN

Parkinson's disease is one of the most common movement disorders and is characterized by dopaminergic cell loss and the accumulation of pathological α-synuclein, but its precise pathogenetic mechanisms remain elusive. To develop disease-modifying therapies for Parkinson's disease, an animal model that recapitulates the pathology and symptoms of the disease, especially in the prodromal stage, is indispensable. As subjects with α-synuclein gene (SNCA) multiplication as well as point mutations develop familial Parkinson's disease and a genome-wide association study in Parkinson's disease has identified SNCA as a risk gene for Parkinson's disease, the increased expression of α-synuclein is closely associated with the aetiology of Parkinson's disease. In this study we generated bacterial artificial chromosome transgenic mice harbouring SNCA and its gene expression regulatory regions in order to maintain the native expression pattern of α-synuclein. Furthermore, to enhance the pathological properties of α-synuclein, we inserted into SNCA an A53T mutation, two single-nucleotide polymorphisms identified in a genome-wide association study in Parkinson's disease and a Rep1 polymorphism, all of which are causal of familial Parkinson's disease or increase the risk of sporadic Parkinson's disease. These A53T SNCA bacterial artificial chromosome transgenic mice showed an expression pattern of human α-synuclein very similar to that of endogenous mouse α-synuclein. They expressed truncated, oligomeric and proteinase K-resistant phosphorylated forms of α-synuclein in the regions that are specifically affected in Parkinson's disease and/or dementia with Lewy bodies, including the olfactory bulb, cerebral cortex, striatum and substantia nigra. Surprisingly, these mice exhibited rapid eye movement (REM) sleep without atonia, which is a key feature of REM sleep behaviour disorder, at as early as 5 months of age. Consistent with this observation, the REM sleep-regulating neuronal populations in the lower brainstem, including the sublaterodorsal tegmental nucleus, nuclei in the ventromedial medullary reticular formation and the pedunculopontine nuclei, expressed phosphorylated α-synuclein. In addition, they also showed hyposmia at 9 months of age, which is consistent with the significant accumulation of phosphorylated α-synuclein in the olfactory bulb. The dopaminergic neurons in the substantia nigra pars compacta degenerated, and their number was decreased in an age-dependent manner by up to 17.1% at 18 months of age compared to wild-type, although the mice did not show any related locomotor dysfunction. In conclusion, we created a novel mouse model of prodromal Parkinson's disease that showed RBD-like behaviour and hyposmia without motor symptoms.


Asunto(s)
Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ratones , Trastornos del Olfato/genética , Enfermedad de Parkinson/genética , Síntomas Prodrómicos , Trastorno de la Conducta del Sueño REM/genética , alfa-Sinucleína/genética , Animales , Recuento de Células , Cromosomas Artificiales Bacterianos , Electroencefalografía , Electromiografía , Endopeptidasa K/metabolismo , Ratones Transgénicos , Trastornos del Olfato/fisiopatología , Enfermedad de Parkinson/fisiopatología , Polimorfismo de Nucleótido Simple , Trastorno de la Conducta del Sueño REM/fisiopatología , Sueño , alfa-Sinucleína/metabolismo
7.
Genes Dev ; 26(10): 1041-54, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22588718

RESUMEN

Autophagy is a lysosomal degradation pathway that converts macromolecules into substrates for energy production during nutrient-scarce conditions such as those encountered in tumor microenvironments. Constitutive mitochondrial uptake of endoplasmic reticulum (ER) Ca²âº mediated by inositol triphosphate receptors (IP3Rs) maintains cellular bioenergetics, thus suppressing autophagy. We show that the ER membrane protein Bax inhibitor-1 (BI-1) promotes autophagy in an IP3R-dependent manner. By reducing steady-state levels of ER Ca²âº via IP3Rs, BI-1 influences mitochondrial bioenergetics, reducing oxygen consumption, impacting cellular ATP levels, and stimulating autophagy. Furthermore, BI-1-deficient mice show reduced basal autophagy, and experimentally reducing BI-1 expression impairs tumor xenograft growth in vivo. BI-1's ability to promote autophagy could be dissociated from its known function as a modulator of IRE1 signaling in the context of ER stress. The results reveal BI-1 as a novel autophagy regulator that bridges Ca²âº signaling between ER and mitochondria, reducing cellular oxygen consumption and contributing to cellular resilience in the face of metabolic stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/inmunología , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Proteínas de la Membrana/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/genética , Línea Celular Tumoral , Endorribonucleasas/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Consumo de Oxígeno , Proteínas Serina-Treonina Quinasas/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Infecciones Estreptocócicas/inmunología , Streptococcus/inmunología , Estrés Fisiológico , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Biol Chem ; 291(27): 14072-14084, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27129202

RESUMEN

B cell lymphoma gene 2 (Bcl-2) family proteins are key regulators of programmed cell death and important targets for drug discovery. Pro-apoptotic and anti-apoptotic Bcl-2 family proteins reciprocally modulate their activities in large part through protein interactions involving a motif known as BH3 (Bcl-2 homology 3). Nur77 is an orphan member of the nuclear receptor family that lacks a BH3 domain but nevertheless binds certain anti-apoptotic Bcl-2 family proteins (Bcl-2, Bfl-1, and Bcl-B), modulating their effects on apoptosis and autophagy. We used a combination of NMR spectroscopy-based methods, mutagenesis, and functional studies to define the interaction site of a Nur77 peptide on anti-apoptotic Bcl-2 family proteins and reveal a novel interaction surface. Nur77 binds adjacent to the BH3 peptide-binding crevice, suggesting the possibility of cross-talk between these discrete binding sites. Mutagenesis of residues lining the identified interaction site on Bcl-B negated the interaction with Nur77 protein in cells and prevented Nur77-mediated modulation of apoptosis and autophagy. The findings establish a new protein interaction site with the potential to modulate the apoptosis and autophagy mechanisms governed by Bcl-2 family proteins.


Asunto(s)
Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Humanos , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/química
9.
J Biol Chem ; 290(44): 26549-61, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26378241

RESUMEN

Autophagy is a catabolic cellular mechanism for entrapping cellular macromolecules and organelles in intracellular vesicles and degrading their contents by fusion with lysosomes. Important roles for autophagy have been elucidated for cell survival during nutrient insufficiency, eradication of intracellular pathogens, and counteracting aging through clearance of senescent proteins and mitochondria. Autophagic vesicles become decorated with LC3, a protein that mediates their fusion with lysosomes. LC3 is a substrate of the cysteine protease ATG4B (Autophagin-1), where cleavage generates a C-terminal glycine required for LC3 conjugation to lipids in autophagosomes. ATG4B both cleaves pro-LC3 and also hydrolyzes lipids from cleaved LC3. We show here that phosphorylation of ATG4B at Ser-383 and Ser-392 increases its hydrolyase activity as measured using LC3 as a substrate. Reconstituting atg4b(-/-) cells with phosphorylation-deficient ATG4B showed a role of ATG4B phosphorylation in LC3 delipidation and autophagic flux, thus demonstrating that the cellular activity of ATG4B is modulated by phosphorylation. Proteolytic conversion of pro-LC3 to LC3-I was not significantly impacted by ATG4B phosphorylation in cells. Phosphorylation-deficient ATG4B also showed reduced interactions with the lipid-conjugated LC3 but not unconjugated LC3. Taken together, these findings demonstrate a role for Ser-383 and Ser-392 phosphorylation of ATG4B in control of autophagy.


Asunto(s)
Autofagia/fisiología , Cisteína Endopeptidasas/metabolismo , Lipoilación/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteolisis , Animales , Proteínas Relacionadas con la Autofagia , Células Cultivadas , Cisteína Endopeptidasas/genética , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Fosforilación/fisiología
10.
Proc Natl Acad Sci U S A ; 110(19): 7808-13, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23603272

RESUMEN

Host innate immune responses to DNA viruses involve members of the nucleotide-binding domain, leucine-rich repeat and pyrin domain containing protein (NLRP) family, which form "inflammasomes" that activate caspase-1, resulting in proteolytic activation of cytokines interleukin (IL)-1ß and IL-18. We hypothesized that DNA viruses would target inflammasomes to overcome host defense. A Vaccinia virus (VACV) B-cell CLL/lymphoma 2 (Bcl-2) homolog, F1L, was demonstrated to bind and inhibit the NLR family member NLRP1 in vitro. Moreover, infection of macrophages in culture with virus lacking F1L (ΔF1L) caused increased caspase-1 activation and IL-1ß secretion compared with wild-type virus. Virulence of ΔF1L virus was attenuated in vivo, causing altered febrile responses, increased proteolytic processing of caspase-1, and more rapid inflammation in lungs of infected mice without affecting cell death or virus replication. Furthermore, we found that a hexapeptide from F1L is necessary and sufficient for inhibiting the NLRP1 inflammasome in vitro, thus identifying a peptidyl motif required for binding and inhibiting NLRP1. The functional importance of this NLRP1-binding motif was further confirmed by studies of recombinant ΔF1L viruses reconstituted either with the wild-type F1L or a F1L mutant that fails to bind NLRP1. Cellular infection with wild-type F1L reconstituted virus-suppressed IL-1ß production, whereas mutant F1L did not. In contrast, both wild-type and mutant versions of F1L equally suppressed apoptosis. In vivo, the NLR nonbinding F1L mutant virus exhibited an attenuated phenotype similar to ΔF1L virus, thus confirming the importance of F1L interactions with NLRP1 for viral pathogenicity in mice. Altogether, these findings reveal a unique viral mechanism for evading host innate immune responses.


Asunto(s)
Regulación Viral de la Expresión Génica , Inmunidad Innata , Inflamasomas/metabolismo , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Animales , Caspasas/metabolismo , Chlorocebus aethiops , Citocinas/metabolismo , Células HEK293 , Células HeLa , Humanos , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Mutación , Fenotipo , Proteínas Recombinantes/metabolismo , Células Vero , Virulencia
11.
J Biol Chem ; 288(18): 12777-90, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23519470

RESUMEN

The cytosolic protein c-FLIP (cellular Fas-associated death domain-like interleukin 1ß-converting enzyme inhibitory protein) is an inhibitor of death receptor-mediated apoptosis that is up-regulated in a variety of cancers, contributing to apoptosis resistance. Several compounds found to restore sensitivity of cancer cells to TRAIL, a TNF family death ligand with promising therapeutic potential, act by targeting c-FLIP ubiquitination and degradation by the proteasome. The generation of reactive oxygen species (ROS) has been implicated in c-FLIP protein degradation. However, the mechanism by which ROS post-transcriptionally regulate c-FLIP protein levels is not well understood. We show here that treatment of prostate cancer PPC-1 cells with the superoxide generators menadione, paraquat, or buthionine sulfoximine down-regulates c-FLIP long (c-FLIP(L)) protein levels, which is prevented by the proteasome inhibitor MG132. Furthermore, pretreatment of PPC-1 cells with a ROS scavenger prevented ubiquitination and loss of c-FLIP(L) protein induced by menadione or paraquat. We identified lysine 167 as a novel ubiquitination site of c-FLIP(L) important for ROS-dependent degradation. We also identified threonine 166 as a novel phosphorylation site and demonstrate that Thr-166 phosphorylation is required for ROS-induced Lys-167 ubiquitination. The mutation of either Thr-166 or Lys-167 was sufficient to stabilize c-FLIP protein levels in PPC-1, HEK293T, and HeLa cancer cells treated with menadione or paraquat. Accordingly, expression of c-FLIP T166A or K167R mutants protected cells from ROS-mediated sensitization to TRAIL-induced cell death. Our findings reveal novel ROS-dependent post-translational modifications of the c-FLIP protein that regulate its stability, thus impacting sensitivity of cancer cells to TRAIL.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Especies Reactivas de Oxígeno/metabolismo , Ubiquitinación , Antimetabolitos/farmacología , Butionina Sulfoximina/farmacología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Inhibidores de Cisteína Proteinasa/farmacología , Regulación hacia Abajo/efectos de los fármacos , Células HEK293 , Células HeLa , Herbicidas/farmacología , Humanos , Leupeptinas , Mutación Missense , Paraquat/farmacología , Fosforilación , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Vitamina K 3/farmacología , Vitaminas/farmacología
12.
Mol Brain ; 17(1): 28, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38790036

RESUMEN

The aggregated alpha-synuclein (αsyn) in oligodendrocytes (OLGs) is one of the pathological hallmarks in multiple system atrophy (MSA). We have previously reported that αsyn accumulates not only in neurons but also in OLGs long after the administration of αsyn preformed fibrils (PFFs) in mice. However, detailed spatial and temporal analysis of oligodendroglial αsyn aggregates was technically difficult due to the background neuronal αsyn aggregates. The aim of this study is to create a novel mouse that easily enables sensitive and specific detection of αsyn aggregates in OLGs and the comparable analysis of the cellular tropism of αsyn aggregates in MSA brains. To this end, we generated transgenic (Tg) mice expressing human αsyn-green fluorescent protein (GFP) fusion proteins in OLGs under the control of the 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) promoter (CNP-SNCAGFP Tg mice). Injection of αsyn PFFs in these mice induced distinct GFP-positive aggregates in the processes of OLGs as early as one month post-inoculation (mpi), and their number and size increased in a centripetal manner. Moreover, MSA-brain homogenates (BH) induced significantly more oligodendroglial αsyn aggregates than neuronal αsyn aggregates compared to DLB-BH in CNP-SNCAGFP Tg mice, suggestive of their potential tropism of αsyn seeds for OLGs. In conclusion, CNP-SNCAGFP Tg mice are useful for studying the development and tropism of αsyn aggregates in OLGs and could contribute to the development of therapeutics targeting αsyn aggregates in OLGs.


Asunto(s)
Cuerpos de Inclusión , Atrofia de Múltiples Sistemas , Oligodendroglía , Agregado de Proteínas , alfa-Sinucleína , Animales , Humanos , Ratones , alfa-Sinucleína/metabolismo , Encéfalo/patología , Encéfalo/metabolismo , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/metabolismo , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Ratones Transgénicos , Atrofia de Múltiples Sistemas/patología , Atrofia de Múltiples Sistemas/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/patología , Agregación Patológica de Proteínas/metabolismo
13.
Neurosci Res ; 163: 43-51, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32145212

RESUMEN

The central nervous system (CNS) uses a significant amount of oxygen for energy production. Decreased oxygen supply due to impaired blood supply critically damages the CNS. As chronic hypoxic conditions have diverse effects via the excessive production of reactive oxygen species, protection from hypoxic damage is important for cell survival. Recent studies have revealed that various markers of hypoxia are altered in age-related neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), indicating the involvement of hypoxia. However, therapeutic strategies targeting hypoxia-induced pathways in ALS have not been developed yet. We previously screened small-molecule compounds that inhibit hypoxia-induced cell death and identified 6-deoxyjacareubin. We hypothesized that the modulation of hypoxia signaling by 6-deoxyjacareubin might protect motor neurons in ALS. Here, we show that 6-deoxyjacareubin indeed ameliorates neurodegeneration in a mouse model of familial ALS. Administration of 6-deoxyjacareubin to this familial ALS model significantly attenuated disease progression and improved locomotor dysfunction. We also found that 6-deoxyjacareubin reduced motor neuron loss and glial activation. Our results indicate that 6-deoxyjacareubin might serve as a potential therapeutic tool for ALS. Moreover, these results suggest that modulation of hypoxia signaling pathways provides a promising strategy to develop therapies for other types of neurodegenerative diseases also characterized by hypoxia.


Asunto(s)
Esclerosis Amiotrófica Lateral , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Animales , Muerte Celular , Modelos Animales de Enfermedad , Hipoxia/complicaciones , Hipoxia/tratamiento farmacológico , Ratones , Ratones Transgénicos , Neuronas Motoras , Piranos , Superóxido Dismutasa , Superóxido Dismutasa-1 , Xantenos
14.
Neurosci Lett ; 765: 136267, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34571089

RESUMEN

For the development of disease-modifying therapies for Parkinson's disease (PD) the identification of biomarkers in the prodromal stage is urgently required. Because PD is considered a systemic disease even in the early stage, we performed a metabolomic analysis of the plasma from a mouse model of prodromal PD (p-PD). Increased levels of isobutyrylcarnitine in p-PD mice imply an abnormality in ß-oxidation in mitochondria, and increased levels of pyrimidine nucleoside can be associated with mitochondrial dysfunction. Consistent with these results, the immunoblot analysis showed a defect in mitochondrial complex I assembly in p-PD mice. These results suggest that systemic mitochondrial dysfunction may exist in p-PD mice and contribute to the pathogenesis of PD, potentially being useful as early biomarkers for PD.


Asunto(s)
Biomarcadores/sangre , Carnitina/análogos & derivados , Mitocondrias/patología , Trastornos Parkinsonianos/metabolismo , Animales , Carnitina/sangre , Modelos Animales de Enfermedad , Metabolómica , Ratones , Mitocondrias/metabolismo , Síntomas Prodrómicos
15.
Neurosci Lett ; 757: 135972, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34033888

RESUMEN

Multiple system atrophy (MSA) is a progressive neurodegenerative disorder characterized by the accumulation of pathogenic phosphorylated α-synuclein in oligodendrocytes. In brains affected by MSA, severe astrogliosis is also observed, but its precise role in MSA pathogenesis remains largely unknown. Recently, the stimulator of interferon genes (STING) pathway and type I interferons, its downstream molecules, have been reported to be involved in the neurodegenerative process and to be activated in astrocytes. This study aimed to investigate the role of the STING pathway in the pathogenesis of MSA using postmortem brains. Samples used for immunohistochemical analysis included 6 cases of MSA parkinsonism type (MSA-P), 6 cases of MSA cerebellar type (MSA-C), and 7 age-matched controls. In MSA-P cases, astrocytes immunopositive for STING and TANK-binding kinase 1 (TBK1), its downstream molecule, were abundantly observed in the putamen and the substantia nigra. Moreover, these molecules colocalized with glial fibrillary acidic protein (GFAP) in reactive astrocytes, and the density of STING-positive astrocytes correlated with that of GFAP-positive reactive astrocytes in the brains of patients with MSA-P. These results suggest that the upregulated expression of STING pathway-related proteins in astrocytes and the subsequent inflammation may contribute to the pathogenesis in MSA-P and could provide novel therapeutic targets for the treatment of MSA.


Asunto(s)
Astrocitos/metabolismo , Proteínas de la Membrana/metabolismo , Atrofia de Múltiples Sistemas/inmunología , Putamen/patología , Sustancia Negra/patología , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Proteína Ácida Fibrilar de la Glía/análisis , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Proteínas de la Membrana/análisis , Persona de Mediana Edad , Atrofia de Múltiples Sistemas/patología , Putamen/citología , Putamen/inmunología , Transducción de Señal/inmunología , Sustancia Negra/citología , Sustancia Negra/inmunología , Regulación hacia Arriba/inmunología
16.
Mol Brain ; 14(1): 80, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33971917

RESUMEN

Homozygous mutations in the lysosomal glucocerebrosidase gene, GBA1, cause Gaucher's disease (GD), while heterozygous mutations in GBA1 are a strong risk factor for Parkinson's disease (PD), whose pathological hallmark is intraneuronal α-synuclein (asyn) aggregates. We previously reported that gba1 knockout (KO) medaka exhibited glucosylceramide accumulation and neuronopathic GD phenotypes, including short lifespan, the dopaminergic and noradrenergic neuronal cell loss, microglial activation, and swimming abnormality, with asyn accumulation in the brains. A recent study reported that deletion of GBA2, non-lysosomal glucocerebrosidase, in a non-neuronopathic GD mouse model rescued its phenotypes. In the present study, we generated gba2 KO medaka and examined the effect of Gba2 deletion on the phenotypes of gba1 KO medaka. The Gba2 deletion in gba1 KO medaka resulted in the exacerbation of glucosylceramide accumulation and no improvement in neuronopathic GD pathological changes, asyn accumulation, or swimming abnormalities. Meanwhile, though gba2 KO medaka did not show any apparent phenotypes, biochemical analysis revealed asyn accumulation in the brains. gba2 KO medaka showed a trend towards an increase in sphingolipids in the brains, which is one of the possible causes of asyn accumulation. In conclusion, this study demonstrated that the deletion of Gba2 does not rescue the pathological changes or behavioral abnormalities of gba1 KO medaka, and GBA2 represents a novel factor affecting asyn accumulation in the brains.


Asunto(s)
Encéfalo/enzimología , Encéfalo/patología , Enfermedad de Gaucher/enzimología , Glucosilceramidasa/metabolismo , Neuronas/enzimología , Neuronas/patología , Oryzias/metabolismo , alfa-Sinucleína/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Autofagia , Enfermedad de Gaucher/patología , Eliminación de Gen , Técnicas de Inactivación de Genes , Modelos Biológicos , Mutación/genética , Fenotipo , Esfingolípidos/metabolismo
17.
FEBS Open Bio ; 10(9): 1758-1764, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32608563

RESUMEN

Ferroptosis, a form of iron-dependent cell death caused by lipid peroxidation, has been implicated in neurological and other disorders. However, the mechanism of ferroptosis in oligodendrocytes is unclear. We tested the susceptibility of MO3.13 cells, an oligodendrocyte line, to ferroptosis after erastin treatment. Immature MO3.13 cells were more susceptible to erastin-induced ferroptosis than chemically differentiated mature MO3.13 cells. Increased expression of solute carrier family 7 member 11 (SLC7A11), which encodes a cystine/glutamate transporter, and greater glutathione concentrations were observed in mature compared with immature MO3.13 cells, linking glutathione to the resistance of mature MO3.13 cells to erastin-induced ferroptosis. These findings highlight the usefulness of immature MO3.13 cells in studies of ferroptosis and investigations into neuropathologies that involve oligodendrocytes.


Asunto(s)
Ferroptosis/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Piperazinas/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos
18.
Apoptosis ; 13(3): 437-47, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18188704

RESUMEN

The HIPPI (HIP-1 protein interactor) protein is a multifunctional protein that is involved in the regulation of apoptosis. The interaction partners of HIPPI include HIP-1 (Huntingtin-interacting protein-1), Apoptin, Homer1c, Rybp/DEDAF, and BAR (bifunctional apoptosis regulator). In search for other binding partners of HIPPI, we performed a yeast two hybrid screen and identified BLOC1S2 (Biogenesis of lysosome-related organelles complex-1 subunit 2) as a novel HIPPI-interacting protein. In co-immunoprecipitation assays, BLOC1S2 specifically associates with HIPPI, but not with HIP-1. To study the expression of BLOC1S2 on the protein level, we generated a mouse monoclonal antibody specific for BLOC1S2 and a multiple tissue array comprising 70 normal and cancer tissue samples of diverse origin. BLOC1S2 protein is widely expressed in normal tissue as well as in malignant tumors with a tendency towards lower expression levels in certain subtypes of tumors. On the subcellular level, BLOC1S2 is expressed in an organellar-like pattern and co-localizes with mitochondria. Over-expression of BLOC1S2 in the presence or absence of HIPPI does not induce apoptosis. However, BLOC1S2 and HIPPI sensitize NCH89 glioblastoma cells to the pro-apoptotic actions of staurosporine and the death ligand TRAIL by enhancing caspase activation, cytochrome c release, and disruption of the mitochondrial membrane potential. Given its interaction with HIPPI and its pro-apoptotic activity, BLOC1S2 might play an important functional role in cancer and neurodegenerative diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Apoptosis/efectos de los fármacos , Glioblastoma/patología , Proteínas/fisiología , Adulto , Secuencia de Aminoácidos , Animales , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Femenino , Células HeLa , Humanos , Inmunohistoquímica , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Datos de Secuencia Molecular , Neoplasias/patología , Unión Proteica , Alineación de Secuencia , Estaurosporina/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Distribución Tisular
19.
FEBS J ; 275(15): 3900-10, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18573101

RESUMEN

Potassium channel tetramerization domain (KCTD) proteins contain a bric-a-brac, tramtrak and broad complex (BTB) domain that is most similar to the tetramerization domain (T1) of voltage-gated potassium channels. Some BTB-domain-containing proteins have been shown recently to participate as substrate-specific adaptors in multimeric cullin E3 ligase reactions by recruiting proteins for ubiquitination and subsequent degradation by the proteasome. Twenty-two KCTD proteins have been found in the human genome, but their functions are largely unknown. In this study, we have characterized KCTD5, a new KCTD protein found in the cytosol of cultured cell lines. The expression of KCTD5 was upregulated post-transcriptionally in peripheral blood lymphocytes stimulated through the T-cell receptor. KCTD5 interacted specifically with cullin3, bound ubiquitinated proteins, and formed oligomers through its BTB domain. Analysis of the interaction with cullin3 showed that, in addition to the BTB domain, some amino acids in the N-terminus of KCTD5 are required for binding to cullin3. These findings suggest that KCTD5 is a substrate-specific adaptor for cullin3-based E3 ligases.


Asunto(s)
Proteínas Cullin/metabolismo , Canales de Potasio/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Cromatografía en Gel , Electroforesis en Gel de Poliacrilamida , Humanos , Microscopía Confocal , Microscopía Fluorescente , Datos de Secuencia Molecular , Canales de Potasio/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
20.
Cancer Res ; 66(18): 9099-107, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16982752

RESUMEN

Hepatitis B virus accounts for more than 1 million cancer deaths annually, but the mechanism by which this virus promotes hepatocellular carcinoma remains unclear. The hepatitis B virus genome encodes an oncoprotein, HBx, which binds various cellular proteins including HBXIP. We show here that HBXIP is a regulator of centrosome duplication, required for bipolar spindle formation in HeLa human carcinoma cells and primary mouse embryonic fibroblast cells. We found that most cells deficient in HBXIP arrest in prometaphase with monopolar spindles whereas HBXIP overexpression causes tripolar or multipolar spindles due to excessive centrosome replication. Additionally, a defect in cytokinesis was seen in HBXIP-deficient HeLa cells, with most cells failing to complete division and succumbing eventually to apoptosis. Expression of viral HBx in HeLa cells mimicked the effects of HBXIP overexpression, causing excessive centrosome replication, resulting in tripolar and multipolar spindles and defective cytokinesis. Immunolocalization and fluorescent protein tagging experiments showed that HBXIP associates with microtubules of dividing cells and colocalizes with HBx on centrosomes. Thus, viral HBx and its cellular target HBXIP regulate centrosome dynamics and cytokinesis affecting genetic stability. In vivo experiments using antisense oligonucleotides targeting HBXIP in a mouse model of liver regeneration showed a requirement for HBXIP for growth and survival of replicating hepatocytes. Thus, HBXIP is a critical regulator of hepatocyte cell growth in vivo, making it a strong candidate for explaining the tumorigenic actions of viral HBx.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Centrosoma/fisiología , Transactivadores/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Procesos de Crecimiento Celular/fisiología , Células HeLa , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Regeneración Hepática , Masculino , Ratones , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Mitosis/fisiología , Huso Acromático/fisiología , Transfección , Proteínas Reguladoras y Accesorias Virales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA